Anda di halaman 1dari 11

ARTHRITIS & RHEUMATISM

Vol. 48, No. 12, December 2003, pp 3464–3474


DOI 10.1002/art.11365
© 2003, American College of Rheumatology

Stem Cell Therapy in a Caprine Model of Osteoarthritis

J. Mary Murphy,1 David J. Fink,1 Ernst B. Hunziker,2 and Frank P. Barry1

Objective. To explore the role that implanted many generations, while retaining their capacity to dif-
mesenchymal stem cells may play in tissue repair or ferentiate when exposed to appropriate signals. The
regeneration of the injured joint, by delivery of an isolation of these cells from adult tissues raises oppor-
autologous preparation of stem cells to caprine knee tunities for the development of novel cellular therapies
joints following induction of osteoarthritis (OA). without the ethical considerations associated with the
Methods. Adult stem cells were isolated from use of embryonic stem cells. Multipotent cells have been
caprine bone marrow, expanded in culture, and trans- isolated from various mesenchymal tissues in adults,
duced to express green fluorescent protein. OA was including skeletal muscle, fat, and synovial membrane
induced unilaterally in the knee joint of donor animals (11–13) as well as hematopoietic (14), neural (15), and
by complete excision of the medial meniscus and resec- hepatic (16) tissues. Because of their multipotentiality
tion of the anterior cruciate ligament. After 6 weeks, a and capacity for self-renewal, adult stem cells may
single dose of 10 million autologous cells suspended in a represent units of active regeneration of tissues dam-
dilute solution of sodium hyaluronan was delivered to aged as a result of trauma or disease (14). In certain
the injured knee by direct intraarticular injection. Con- degenerative diseases such as osteoarthritis (OA), stem
trol animals received sodium hyaluronan alone. cells are depleted and have reduced proliferative capac-
Results. In cell-treated joints, there was evidence ity and reduced ability to differentiate (17). The systemic
of marked regeneration of the medial meniscus, and or local delivery of stem cells to these individuals may
implanted cells were detected in the newly formed therefore enhance repair or inhibit the progressive loss
tissue. Degeneration of the articular cartilage, osteo- of joint tissue.
phytic remodeling, and subchondral sclerosis were re- OA is characterized by degeneration of the artic-
duced in cell-treated joints compared with joints treated
ular cartilage, with loss of matrix, fibrillation, formation
with vehicle alone without cells. There was no evidence
of fissures, and ultimately complete loss of the cartilage
of repair of the ligament in any of the joints.
surface. Other articular tissues are also affected, includ-
Conclusion. Local delivery of adult mesenchymal
ing the subchondral bone, ligaments, joint capsule, syno-
stem cells to injured joints stimulates regeneration of
vial membrane, and periarticular muscles (18,19). Al-
meniscal tissue and retards the progressive destruction
though OA affects a large proportion of the population,
normally seen in this model of OA.
there are few, if any, effective therapies available today
that alter the pathobiologic course of the disease (20).
Mesenchymal stem cells (MSCs) have the capac-
The objective of this study was to determine if
ity to differentiate into a variety of connective tissue
delivery of stem cells to the knee following traumatic
cells (1–4) including bone, cartilage, tendon, muscle, and
injury would enhance repair of damaged tissue or im-
adipose tissue (3,5–10). These cells may be isolated from
pede the progression to OA that is the usual conse-
bone marrow with ease and expanded in culture through
quence of such injuries (21). There are several examples
of the use of stem cells for articular cartilage repair
1
J. Mary Murphy, PhD, David J. Fink, PhD, Frank P. Barry, (22,23). There are also reports describing the use of
PhD: Osiris Therapeutics, Baltimore, Maryland; 2Ernst B. Hunziker, these cells for the repair of segmental bone defects in
MD: M. E. Müller Institute for Biomechanics, University of Bern,
Bern, Switzerland. long bones (24). However, there are no reports describ-
Address correspondence and reprint requests to Frank P. ing the use of these cellular therapies in trauma-induced
Barry, PhD, Osiris Therapeutics Inc., 2001 Aliceanna Street, Balti- OA. In general, the cells have been delivered to either
more, MD 21231. E-mail: fbarry@osiristx.com.
Submitted for publication February 4, 2003; accepted in cartilage or bone using a 3-dimensional scaffold that is
revised form August 28, 2003. fixed to the defect site, usually by means of an open
3464
STEM CELL THERAPY IN OA 3465

surgical procedure. There are many issues associated Retroviral vector construction and virus production.
with the selection of the scaffold material, including its Caprine MSCs were transduced to express the Aequorea
Victoria enhanced green fluorescent protein (eGFP) using the
ability to support cell viability and differentiation and its
retroviral vector pOT24. The construction of this vector has
retention and degradation in situ. been described previously (29). Briefly, the GFP-1 gene from
In the present study, we used a simpler, scaffold- pEGFP-1 (Clontech, Palo Alto, CA) was cloned into the
free approach in which the cells were delivered as a pJM573neo retroviral vector (30,31). Vector supernatants con-
suspension by direct intraarticular injection. The suspen- taining different pseudotyped vector particles were generated
using the gibbon ape leukemia virus envelope containing the
sion was prepared in a dilute solution of sodium hyalu- PG13 packaging cell line (32). Retroviral supernatants con-
ronan, which is commonly used for the treatment of OA taining the GFP vector were produced as follows: plasmid
(25) and also has the effect of increasing the chondro- pOT24 was transfected into GP&E86 ecotropic producer cells
genic activity of MSCs (26). There was evidence of cell (American Type Culture Collection [ATCC] CRL-9642) (33)
using DOTAP (Boehringer-Mannheim, Indianapolis, IN) as
engraftment in several tissues in the joint and a marked
directed by the manufacturer. The transfected cells were grown
regeneration of the excised meniscus. In addition, there at 37°C, 5% CO2, 90% humidity in DMEM–high glucose
was evidence of a reduction in OA progression in the medium supplemented with 10% heat-inactivated FBS, 1%
cell-treated joints. We suggest that the enhanced repair penicillin–streptomycin, and 0.5 mg/ml protamine sulfate–
response results from interaction between the implanted G418 (Sigma) as a selective agent.
Cultures were grown to 70% confluence, the medium
cells and fibroblasts derived from the host synovium at was replaced with fresh retroviral medium (without G418), and
the site of injury. the cells were incubated at 32°C for 2 days. The ecotropic
retrovirus was used to transduce the gibbon ape leukemia virus
envelope containing the PG13 retroviral producer cells (ATCC
MATERIALS AND METHODS CRL-9078). Transduction was performed using the centrifugal
transduction procedure outlined below, followed by selection
Preparation of caprine MSCs. A heparinized bone
with G418 (0.5 mg/ml). The culture medium containing the
marrow aspirate (3–7 ml) was obtained from the iliac crest of
retroviral vectors was collected, filtered through a 0.45-␮m
castrated male Western Cross goats. The aspirate was washed
filter, and stored at ⫺80°C.
with medium (Dulbecco’s modified Eagle’s medium Transduction and expansion of caprine MSCs. Pri-
[DMEM]–low glucose; Hyclone, Logan, UT) containing 1% mary frozen cultures of caprine MSCs were thawed and plated
antibiotic–antimycotic (Gibco, Grand Island, NY), and centri- in T-75 flasks at 500,000 cells/flask with DMEM–high glucose
fuged. The precipitated cells were then suspended in medium containing 1% antibiotic–antimycotic and 10% FBS. After 1
with 10% fetal bovine serum (FBS; Hyclone) at a final density day, the culture medium was aspirated and 15 ml of retroviral
of 1.4–1.6 ⫻ 106 cells/ml. Cells were seeded on T-185 flasks supernatant, containing 8 mg/ml polybrene (Sigma), was added
and maintained at 37°C with 95% humidity and 5% CO2 in the to each flask. Culture medium with polybrene only was used
same medium. After 5 days, red blood cells were washed off for mock-transduced cultures. Following transduction for 24
with phosphate buffered saline (PBS) and fresh medium was hours, the vector-containing medium was aspirated and a
added. Colonies of adherent cells formed within 9 days and the second round of transduction was performed with fresh retro-
colonies were trypsinized from the flasks when the colonies viral supernatant. Selection of transduced cells with G418 (1.0
covered 60–90% of the plate. The cells were cryopreserved at mg/ml) was performed for 7–10 days until confluence was
the end of primary culture. reached. The selected cells were trypsinized and expanded to
Differentiation assays. Chondrogenesis was performed the end of passage 2. A sample of the cells was washed with
with modifications of the pellet culture system described PBS containing 2% weight/volume bovine serum albumin and
previously (7,9), using dexamethasone at 10 nM (Sigma, St. 0.1% w/v sodium azide for assessment of eGFP using
Louis, MO) and 10 ng/ml recombinant human transforming fluorescence-activated cell sorter (FACS) analysis. Intrinsic
growth factor ␤3 (Oncogene Research Products, San Diego, fluorescence from the GFP-transduced MSCs was compared
CA). After 14 days in culture, pellets were harvested and fixed with that from untransduced controls. All cells were resus-
in 10% formalin, and cut sections were stained with Safranin O pended in FACS buffer containing 1% paraformaldehyde
and with an antibody specific for type II collagen (27). For (Electron Microscopy Sciences, Fort Washington, PA) imme-
osteogenic differentiation, the cells were cultured in 10 nM diately before analysis. Cells were analyzed by collecting 10,000
dexamethasone, 10 nM ␤-glycerophosphate, 50 ␮M ascorbic events on a Becton-Dickinson Vantage instrument using
acid 2-phosphate, 100 nM prostaglandin E2, and 5% FBS CellQuest software (Becton-Dickinson, Franklin Lakes, NJ).
(6,28). To evaluate the differentiation potential of the cells in Surgical protocol. The Institutional Animal Care and
ectopically implanted scaffolds, MSCs were loaded onto Use Committee approved all procedures used. Castrated male
hydroxyapatite/tricalcium phosphate cubes (3 ⫻ 3 ⫻ 3 mm; Western Cross goats (weighing 70–112 kg, ages 29–34 months;
Zimmer, Warsaw, IN) at a density of 5 ⫻ 106 cells/ml, and n ⫽ 24), confirmed to be free of Q fever, brucellosis, and caprine
implanted subcutaneously on the dorsal surface of nude mice arthritis encephalitis, were used. Animals were treated with
(5). The cubes were removed 6 weeks later and sections were butorphenol (0.05–0.10 mg/kg) and diazepam (0.1–0.2 mg/kg) by
stained with hematoxylin and modified Mallory aniline blue for intravenous injection and with an equal mixture of ketamine (100
the presence of bone and cartilage. mg/ml) and diazepam (5 gm/ml) at a dose of 0.5 ml/kg as
3466 MURPHY ET AL

Table 1. Study design*


Postsurgery
termination,
Group No. Treatment weeks
1 (control) 3 Vehicle injection 6 weeks postsurgery 12
2 (test) 6 GFP-transduced MSC/vehicle injection 12
6 weeks postsurgery
3 (control) 6 Vehicle injection 6 weeks postsurgery 26
4 (test) 9 GFP-transduced MSC/vehicle injection 26
6 weeks postsurgery

* GFP ⫽ green fluorescent protein; MSC ⫽ mesenchymal stem cell.

anesthetic. Intraoperative anesthesia was maintained with isoflu-


rane (⬃1.5%) in oxygen delivered by endotracheal tube. Figure 1. Study design. Experimental plan used to evaluate the effect
For combined anterior cruciate ligament (ACL) tran- of delivery of autologous stem cells to the knee following meniscec-
section and medial meniscectomy, a lateral parapatellar skin tomy and resection of the anterior cruciate ligament (ACL) for
incision was made beginning at a level 2 cm proximal to the induction of osteoarthritis. Color figure can be viewed in the online
patella and extending to the level of the tibial plateau. Subcu- issue, which is available at http://www.arthritisrheum.org.
taneous tissue was incised, and the lateral fascia was separated
from the joint capsule for ⬃1 cm in either direction away from
the incision. The lateral aspect of the vastus lateralis and the pension was injected into the medial compartment of the
joint capsule were incised and the patella was luxated medially operated joint after aspiration of synovial fluid. An 18-gauge
to expose the trochlear groove and medial and lateral condyles needle was inserted posterior to the medial edge of the patellar
of the distal femur. ligament, through the triangle formed by the epicondyle of the
ACL removal was performed by first excising its at- femur, the meniscal/tibial plateau, and the notch formed by
tachment on the medial aspect of the lateral femoral condyle. their junction. Following injection, the joint was repeatedly
The proximal attachment was brought forward and the entire flexed and extended for dispersal of the suspension throughout
ligament was excised from its tibial attachment. The stifle was the intraarticular space.
moved in a drawer test to ensure that the entire cruciate Histochemical analysis of bone and cartilage. Imme-
ligament had been excised. The medial meniscus was removed diately after the animals were killed, the distal head of the
by sharp excision. The caudal horn of the meniscus was femur and the proximal tibial plateau were removed and fixed
grasped with a hemostat and its axial (lateral) attachment was in 10% formalin. The medial condyles and the anterior portion
excised from its tibial attachment. Working from caudal to of the tibial plateau were dehydrated through a series of
lateral, then cranial, the meniscus was excised from its attach- increasing ethanol concentrations and embedded in methyl
ments until it was completely removed. ACL excision was methacrylate. Serial sagittal sections were cut using a diamond
carried out first, followed by meniscectomy.
Study design. Animals were randomized into 4 groups
as described in Table 1 and Figure 1. The control groups were Table 2. Histologic grading scheme
not different from the test groups with respect to age and
weight, but the test animals underwent a bone marrow aspira- Parameter, grade Description
tion for cell preparation 2 weeks prior to surgery. OA was Articular cartilage structure
induced unilaterally in the knee joint of donor animals by 0–10 Ref. 35*
complete excision of the medial meniscus and resection of the Reduction of articular
ACL as described above, and after a recovery period of 3 cartilage matrix staining
weeks, all animals were exercised once daily by having them 0 None
run on a hard surface for a distance of 90 meters. At all other 1 Mild
times, animals were allowed free movement in an unconfined 2 Moderate
3 Severe
environment. Control animals received an injection of vehicle Presence of osteophytes
alone in the operated joint 6 weeks after surgery. This con- 0 None
sisted of 5 ml sodium hyaluronan (Hylartin-V; Pharmacia & 1 Cartilage/connective tissue
Upjohn, Peapack, NJ) at a concentration of 4 mg/ml. Test 2 Mainly cartilage/some bone formation
animals received a single intraarticular injection of 10 ⫻ 106 3 Mainly bone formation
autologous GFP-transduced MSCs as a suspension in the Subchondral bone plate
vehicle at 6 weeks after surgery. thickening
Intraarticular injection of MSCs. Frozen cells were 0 None
thawed rapidly at 37°C, washed with culture medium and PBS, 1 Mild
2 Moderate
centrifuged, and resuspended in Hylartin solution at a density 3 Severe
of 2 ⫻ 106 cells/ml. Goats were anesthetized, intubated, and
placed in dorsal recumbency. Five milliliters of the cell sus- * 0 ⫽ normal; 10 ⫽ complete loss to subchondral bone.
STEM CELL THERAPY IN OA 3467

Figure 2. Isolation and characterization of mesenchymal stem cells (MSCs) from goat marrow.
Adherent cells from bone marrow aspirates grew as a monolayer in passage 1 culture (A) (original
magnification ⫻ 40) and were uniformly SH4⫹/CD14⫺ by fluorescence-activated cell sorter
analysis (B). Goat MSCs deposited calcium in osteogenic cultures in vitro (C) and were capable of
elaborating a proteoglycan-rich (D) and type II collagen–positive (E) extracellular matrix in
chondrogenic pellet cultures in vitro. Goat MSCs loaded on a hydroxyapatite/tricalcium phosphate
scaffold (S) and implanted subcutaneously in nude SCID mice showed differentiation into bone
(B), fat (L), marrow (M), and cartilage (C) after 6 weeks in vivo (F and G), while control, unloaded
scaffolds showed no evidence of mesenchymal differentiation and the pores (P) were empty or
infiltrated with host hematopoietic cells (H). Bar ⫽ 100 ␮M.

saw (Leco, St. Joseph, MN) from the tissues of both the Sulfated glycosaminoglycan was visualized by staining with
operated and contralateral (unoperated) joints. The sections toluidine blue for 5 minutes at 60°C and with 0.1% Safranin O
were glued onto plexiglass object holders, milled to a thickness for 5 minutes at room temperature. Type I collagen was
of ⬃100 ␮m with a Polycut E apparatus (Reichert-Jung, detected using monoclonal antibody I-8H5 at 0.2 ␮g/ml (On-
Nussloch, Germany), and polished and surface-stained with cogene, San Diego, CA) and type II collagen using monoclonal
McNeil’s tetrachrome/toluidine blue O/basic fuchsine (34). antibody C4F6 (27) at 0.5 ␮g/ml. Sections were stained using
Sections from each condyle and tibial plateau (a minimum of the EnvisionSystem autostainer (Dako, Carpinteria, CA). In-
3) were graded by a blinded assessor based on the following cubation with the primary antibody was for 30 minutes.
parameters: 1) articular cartilage structure, 2) reduction of Visualization was performed with diaminobenzidine and coun-
articular cartilage matrix staining, 3) presence of osteophytes, terstaining with hematoxylin.
and 4) subchondral bone plate thickening. The articular carti- Statistical analysis. Comparisons between the histo-
lage structure was graded on a scale ranging from 0 (normal) logic scores for each group were made using analysis of
to 10 (complete loss to subchondral bone) as described previ- variance. A P value of less than 0.05 was significant.
ously (35). The remaining parameters were scored 0–3 (as
described in Table 2). RESULTS
Histochemical analysis of meniscal tissue. Neomenis-
cal tissue from the posterior horn in the medial compartment Characterization of caprine MSCs. MSCs were
was fixed in 10% buffered formalin and embedded in paraffin. isolated from bone marrow aspirates obtained from the
3468 MURPHY ET AL

superior iliac crest of adult male goats and expanded to


form confluent cultures of adherent cells with a fibro-
blastic morphology (Figure 2A). The cells were homo-
geneously SH4⫹/CD14⫺ (Figure 2B) and CD44⫹/
CD34⫺ (3). The capacity of the cells to differentiate
into chondrocytes and osteocytes was demonstrated in
vitro (Figures 2C–E), and in vivo when implanted sub-
cutaneously in nude mice (Figures 2F and G). The
efficiency of transduction was 62.3 ⫾ 6.5% (mean ⫾
SD). Transduction of human MSCs with eGFP had no
effect on the differentiation potential of the cells, and
transgene expression was maintained after induction
along the osteogenic, adipogenic, and chondrogenic
pathways (36).
Clinical observations. In this study we assessed
the capacity of MSCs to impact OA progression in goats Figure 3. Appearance of regenerated medial meniscus after intraar-
ticular treatment with autologous mesenchymal stem cells (MSCs).
following trauma to the joint caused by unilateral medial The cell treatment resulted in the formation of tissue in the posterior
meniscectomy and transection of the ACL. All animals compartment at 12 weeks after complete medial meniscectomy. The
were mobile 1–2 hours after surgery and there were no excised tissues (B and C) inserted between the distal head of the femur
instances of infection. No immobilization or splinting of and proximal tibial plateau (A) are shown. Regenerated hyaline-like
the joints was used and animals were bearing weight on meniscal tissue is also evident at 20 weeks after MSC injection (D and
E). The regenerated meniscal horn protected the posterior tibial
the operated joint within 3–5 days. Animals tolerated the plateau (D) and had reformed a functional enthesis with the tibial bone
cell injection well, and there was no evidence of local via the meniscal insertional ligament (arrow in E).
inflammation, immobilization, or unloading of the joint
resulting from the cell treatment.
The effect of delivery of autologous GFP- of disorganized repair tissue at the same site, which was
transduced MSCs on the joint pathology was assessed by generally attached to the proximal tibia, and there was
macroscopic evaluation. Appearance of meniscal-like little evidence of extension between the articulating
repair tissue, or neomeniscus, was observed in associa- surfaces (result not shown). There was no evidence of
tion with the posterior medial compartment of cell- repair of the severed ACL in any of the joints, regardless
treated knees at 6 weeks after injection (Figures 3A–C). of treatment.
This tissue was hyaline in nature and appeared to Protection from OA damage. Transection of the
provide a bearing surface for the tibial and femoral ACL and complete medial meniscectomy in the caprine
condyles. Immunohistochemical staining of the neome- knee resulted in the development of lesions characteris-
niscus indicated a dense, type I collagen–containing tic of OA (37–41), including 1) large areas of erosion of
network surrounding cells with fibroblastic morphology the articular cartilage on the femoral condyle and tibial
and some areas containing cells of rounded morphology plateau, 2) formation of periarticular osteophytes, and
surrounded by a type II collagen–positive matrix (results 3) changes to the trabecular organization of the sub-
not shown). There was also evidence of vascularization chondral bone. We carried out a histologic assessment of
in the neomeniscus close to the point of synovial attach- the medial femoral condyle from all animals to deter-
ment. This tissue was not observed in control mine if there were differences between the cell-treated
(hyaluronan-treated) animals at this time point. and control groups. In the control (vehicle-treated)
Twenty weeks after delivery of cells there was joints, there was substantial fibrillation of the articular
again evidence of repair tissue associated with the surface with loss of extracellular matrix, as well as large
posterior medial compartment in 7 of 9 treated joints. areas of osteophytic remodeling (Figures 4A–F) as
The tissue was organized, generally detached from both compared with the contralateral (unoperated) joints
femoral and tibial surfaces, and extended into the artic- (Figure 4Q). In the cell-treated joints, the degree of
ulating region between these areas (Figures 3D and E). cartilage destruction, osteophyte formation, and sub-
Functional entheses with the tibial bone were formed chondral sclerosis were all reduced compared with that
with repair of the meniscal insertional ligaments (Figure in the control joints (Figures 4G–N). In 4 of the 6
3E, arrow). Control joints at this time showed evidence cell-treated joints (Figures 4G–N), there was less dam-
STEM CELL THERAPY IN OA 3469

Figure 4. Microscopic analysis of the medial femoral condyle. Goat knee joints were subjected to total medial meniscectomy and anterior cruciate
ligament resection, which was followed 6 weeks later by an intraarticular injection of hyaluronan (A–F) or mesenchymal stem cells (MSCs)
resuspended in hyaluronan (G–P). Osteoarthritic changes such as proteoglycan depletion (indicated by a reduction in surface staining), severe
fibrillation, and loss of cartilage, and osteophytes (Os) and bone remodeling were evident in vehicle-treated joints (A–E). Large chondrocyte clones
(arrows) were evident in areas distant to the primary lesion (F). Boxed areas in A, C, and E are shown as expanded images in B, D, and F,
respectively. In joints that demonstrated evidence of meniscal regeneration after MSC application (G–N), changes to the cartilage and bone were
much less severe. Mild surface roughening (H) and proteoglycan depletion in the surface zone (H–N) were evident. Proliferation of cells at the
cartilage surface was also seen (L and N). Boxed areas in G, I, K, and M are shown as expanded images in H, J, L, and N, respectively. Protection
from severe osteoarthritic changes was not evident in cell-treated joints in areas where meniscal regeneration did not occur. In a contralateral
(unoperated) control joint from the same goat as in E, the appearance of a normal condyle can be seen (Q). Bar ⫽ 1 mm.
3470 MURPHY ET AL

rounded morphology that were type II collagen positive


(Figures 6E and F). Twenty weeks after injection, the
regenerated meniscus had large areas positive for pro-
teoglycan and type II collagen with the typical appear-
ance of fibrocartilage (Figures 6G–J).
Cell engraftment in the injured joint was evalu-
ated in a separate experiment by delivery of GFP-
transduced cells by intraarticular injection into goat
knee joints 12 weeks following ACL resection or com-
plete medial meniscectomy. Cells were also injected into
the noninjured contralateral joints. Seven days after
injection, labeled cells were detected in the synovial fluid
Figure 5. Histologic grading of the joint. Changes to the middle and synovial fluid lavage whether or not OA was
medial condyle after 6 weeks of exposure to the mesenchymal stem
cells (MSC) were independently assessed with regard to articular
present. These cells were viable and proliferated when
cartilage structure, reduction of articular cartilage matrix staining, the seeded in tissue culture flasks (results not shown). In
presence of osteophytes, and subchondral bone plate thickening. addition, the injected cells colonized and integrated into
Scores were compared using analysis of variance. ⴱ ⫽ P ⬍ 0.05 surface layers of soft tissues within the joint, including
between groups. Bars show the mean and SD. the synovial lining, fat pad, and lateral meniscus. In
some of these experiments, sodium hyaluronan was used
as the vehicle and, in other cases, PBS was used, but the
age to the articular surface and less evidence of osteo- nature of the vehicle had no effect on cell engraftment.
phytic changes, indicating that there was some degree of Likewise, the pattern of distribution of injected cells was
protection provided to the condyle. In the remaining 2 the same whether ACL transection or meniscectomy was
cell-treated joints (Figures 4O and P), there was less used.
evidence of protection of the condyle. In these joints, The synovial capsule, fat pad, and lateral menis-
there was little evidence of formation of neomeniscus. cus showed a high incidence of cell engraftment (Figures
Reduction of articular cartilage matrix staining, 7A and B). In some cases, cell engraftment was detected
changes in osteophyte formation, and subchondral bone on the epiligament, or sheath, of the posterior cruciate
plate thickening were graded as described in Table 2 ligament and the extensor digitorum longus. Analysis of
(35). Histologic scores for all parameters were closer to articular cartilage from the middle regions of the medial
normal in the middle medial condyle of cell-treated and lateral femoral condyles and from the medial and
joints 6 weeks after injection, and treatment had a lateral unprotected tibial plateaus showed no cell en-
significant effect on maintenance of the articular carti- graftment on either the intact or the fibrillated cartilage.
lage structure and subchondral bone plate thickening Small, fluorescently bright bodies were found that were
(Figure 5). Twenty weeks after cell injection, there were similar in size and fluorescence intensity to red blood
significant OA lesions in both the cell-treated and cells. GFP-positive cells were found in some areas of
control joints, despite the presence of neomeniscal tissue synovial lining on both the intracondylar aspect and the
in the treated joints. It is likely that the cumulative effect outer aspect of the lateral and medial condyles (Figure
of the abnormal load imposed as a result of the severed 7C).
ACL resulted in progressive cartilage damage that was
not prevented by repair of the meniscus alone.
DISCUSSION
MSC engraftment. Fluorescence microscopy of
sections of neomeniscal tissue indicated that the GFP- This study evaluates the utility of stem cell ther-
transduced implanted cells were associated with the apy for delaying the progression of arthritic lesions that
regenerated tissue (Figures 6A–D). The cells were gen- occur following joint injury. Adult MSCs were delivered
erally associated with the surface (Figures 6B and C) of by intraarticular injection as a suspension without the
the tissue and, in some cases, were also detected in the use of a solid biomatrix 6 weeks after total medial
interior (Figure 6D). Immunohistochemical staining in- meniscectomy and resection of the ACL. The involve-
dicated a dense, type I collagen–rich fibrillar network ment of injected GFP-transduced cells with the devel-
populated with cells with a fibroblastic morphology. In opment of appreciable neomeniscal tissue in treated
addition, there were regions containing cells with a joints was associated with protection against degenera-
STEM CELL THERAPY IN OA 3471

Figure 6. Microscopic analysis of regenerated meniscal tissue. Green fluorescent protein (GFP)–positive
cells were detected primarily at the surface (B and C) and also in the center (D) of neomeniscal tissue 6
weeks after injection of GFP-transduced mesenchymal stem cells. Neomeniscal tissue not exposed to the
joint environment was used as a negative control (A). Immunohistochemical staining of the posterior
meniscal-like tissue indicated a dense, cellular, type I collagen positive fibrous network (results not shown)
with small areas of more rounded cells that were type II collagen positive (E and F). By 20 weeks after
injection, the neomeniscus had areas of Safranin O–positive proteoglycan and type II collagen (G and I,
respectively) in a type I collagen background (H). Further analysis of the type II collagen staining (boxed
area of I) showed the typical appearance of fibrocartilage (J). (Original magnification ⫻ 200 in A–D; ⫻
20 in G–I; ⫻ 100 in E, F, and J.)

tive changes in these cell-treated joints 6 weeks after with intact ligaments (21). Ligament rupture associated
treatment. The biphasic ultrastructure of meniscus and with meniscal injury further increases this risk, and the
articular cartilage is critical for load distribution, a fact that reconstruction of the ACL was not performed
smooth low-friction gliding surface, and resilience to in this study may explain the continuing degradation of
compression in the complex biomechanics of the knee treated joints that occurred at the 26-week time point.
joint (42). Data on the incidence of OA after meniscal Several surgical and tissue-engineering ap-
injury highlight the importance of restoring a functional proaches are currently in use for the repair of meniscal
meniscus as soon as possible after injury. Even after tissue. These have involved the use of a broad variety of
repair, isolated meniscal rupture has been associated materials, including small intestine submucosa (43),
with a 10-fold increase in the risk of development of devitalized tissue (44) or collagen scaffolds (45), or the
arthrosis, and meniscectomy doubles this risk in a joint placement of meniscal allograft tissue derived from
cadavers (46,47). These approaches are specifically de-
signed for replacement of damaged meniscal tissue but
may have no general effect on surrounding joint tissues.
Human and animal studies have shown a regenerative
response to meniscectomy (48–50). This regeneration,
although minimal, is enhanced in the case of human
studies by preservation of the meniscal rim (50). MSCs
Figure 7. Retention of cells in the joint. Green fluorescent protein– may contribute to and enhance this normal repair pro-
transduced mesenchymal stem cells, injected into normal (noninjured) cess.
joints or into joints 12 weeks after medial meniscectomy or anterior Some augmentation of natural repair by MSCs
cruciate ligament resection, were found to be integrated into the cell
layers lining joint structures such as the meniscus (A), synovial capsule
loaded on a collagen sponge in a rabbit partial menis-
(B), and periosteum on the medial aspect of the medial condyle (C) 1 cectomy model has been described (51). Activation of
week after injection. mesenchymal cells is an important and perhaps rate-
3472 MURPHY ET AL

limiting step in the formation of granulation tissue cell delivery and is therefore unencumbered by the
during wound healing (52). Hyaluronan also contributes complexities associated with placement of a solid cell
to the granulation phase of both fetal and adult wound construct. Other cell-based approaches are possible that
healing (53,54) and stimulates the migration and mitosis might enhance the effects observed in this study. For
of mesenchymal and epithelial cells (55,56). MSC-based example, direct implantation of meniscal cells, chondro-
repair in the presence of hyaluronan may therefore cytes, or stem cells, perhaps in combination with delivery
accelerate and amplify the natural repair process of of appropriate mitogens or growth factors to stimulate
recruiting these cells to the site of tissue repair or host cell proliferation, may lead to a sustained therapeu-
regeneration, and may contribute to the formation of tic effect. Alternative approaches might also involve the
new meniscus after meniscectomy. Meniscal regenera- delivery of cell-binding or cytotactic factors to enhance
tion after partial meniscectomy in a rabbit model was the local progenitor cell population, leading ultimately
enhanced by 5 injections of hyaluronan (57). Because to the reversal of the degradative process. Finally,
the cells used in the present study were retrovirally because of the close integration of the implanted cells
transduced to express GFP, it is conceivable that ex- with host tissues, use of genetically modified cells to
pressed GFP or the vector used for the transduction may deliver therapeutic genes to the site of injury in the joint
have affected the outcome. However, transduction of may also enhance the observed effects.
the cells did not affect their capacity to proliferate, and
engraftment of the transduced cells in the neomeniscus
occurred without evidence of an immune response at ACKNOWLEDGMENTS
this site or elsewhere in the joint. We thank Stephanie Oppenheimer, Jerry Skwarek,
Several tissue-engineering approaches have been Mike Ponticiello, Karl Kavalkovich, and Don Simonetti for
used for the repair of joint lesions. For example, the their expert technical assistance, and Drs. Alastair Mackay and
Arnold Caplan for critical review of the manuscript.
fixation of implanted chondrocytes beneath a sutured
flap of ectopic tissue, such as periosteum, has been
widely used for the treatment of cartilage defects (58). REFERENCES
Other approaches have centered on the use of cells 1. Friedenstein AJ. Precursor cells of mechanocytes. Int Rev Cytol
loaded on a scaffold and delivered to a lesion site. These 1976;47:327–59.
methods are applicable to the repair of focal defects of 2. Owen M, Friedenstein AJ. Stromal stem cells: marrow-derived
osteogenic precursors. In: Evered D, Hartnett S, editors. Cell and
defined dimensions, but not to the treatment of complex molecular biology of vertebrate hard tissues. Chichester, UK:
lesions that cover a large surface area of the joint and Wiley; 1988. p.42–60.
that may be associated with severe and progressive 3. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R,
Mosca JD, et al. Multilineage potential of adult human mesenchy-
inflammatory conditions such as OA. Multipotent cells mal stem cells. Science 1999;284:143–7.
have been isolated from the surface zone of articular 4. Colter DC, Sekiya I, Prockop DJ. Identification of a subpopulation
cartilage (59) and MSCs have the capacity to repair of rapidly self-renewing and multipotential adult stem cells in
colonies of human marrow stromal cells. Proc Natl Acad Sci U S
fibrillated cartilage in vitro (60). It is attractive to A 2001;98:7841–5.
hypothesize that MSCs could have a role in cartilage 5. Haynesworth SE, Goshima J, Goldberg VM, Caplan AI. Charac-
protection by a direct resurfacing of the articular carti- terization of cells with osteogenic potential from human marrow.
Bone 1992;13:81–8.
lage or act to preserve subchondral or trabecular bone 6. Jaiswal N, Haynesworth SE, Caplan AI, Bruder SP. Osteogenic
structure–associated mechanical integrity of the joint. differentiation of purified, culture-expanded human mesenchymal
Early bone changes have been associated with develop- stem cells in vitro. J Cell Biochem 1997;64:295–312.
7. Mackay AM, Beck SC, Murphy JM, Barry FP, Chichester CO,
ment of OA and may precede changes to the articular Pittenger MF. Chondrogenic differentiation of cultured human
cartilage (61,62). MSCs injected into the knee joint did mesenchymal stem cells from marrow. Tissue Engineering 1998;4:
not bind to normal or fibrillated articular cartilage in 415–28.
8. Young RG, Butler DL, Weberm W, Caplan AI, Gordon SL, Fink
vivo, but we cannot exclude the hypothesis that the cells DJ. Use of mesenchymal stem cells in a collagen matrix for
may act to mediate OA progression by an effect other Achilles tendon repair. J Orthop Res 1998;16:406–13.
than that seen on meniscal regeneration. 9. Barry F, Boynton RE, Liu B, Murphy JM. Chondrogenic differ-
entiation of mesenchymal stem cells from bone marrow: differen-
This study suggests that there may be a therapeu- tiation-dependent gene expression of matrix components. Exp Cell
tic benefit associated with intraarticular injection of Res 2001;268:189–200.
stem cells following traumatic injury to the knee. The 10. Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human
mesenchymal stem cells differentiate to a cardiomyocyte pheno-
longer term effect of this may be a reduction or delay in type in the adult murine heart. Circulation 2002;105:93–8.
the progression to OA. This is a scaffold-free method for 11. De Bari C, Dell’Accio F, Tylzanowski P, Luyten FP. Multipotent
STEM CELL THERAPY IN OA 3473

mesenchymal stem cells from adult human synovial membrane. 34. Hunziker EB. Growth-factor-induced healing of partial-thickness
Arthritis Rheum 2001;44:1928–42. defects in adult articular cartilage. Osteoarthritis Cartilage 2001;
12. Gronthos S, Franklin DM, Leddy HA, Robey PG, Storms RW, 9:22–32.
Gimble JM. Surface protein characterization of human adipose 35. Carlson CS, Guilak F, Vail TP, Gardin JF, Kraus VB. Synovial
tissue-derived stromal cells. J Cell Physiol 2001;189:54–63. fluid biomarker levels predict articular cartilage damage following
13. Deasy BM, Jankowski RJ, Huard J. Muscle-derived stem cells: complete medial meniscectomy in the canine knee. J Orthop Res
characterization and potential for cell-mediated therapy. Blood 2002;20:92–100.
Cells Mol Dis 2001;27:924–33. 36. Lee K, Majumdar MK, Buyaner D, Hendricks JK, Pittenger MF,
14. Weissman IL. Translating stem and progenitor cell biology to the Mosca JD. Human mesenchymal stem cells maintain transgene
clinic: barriers and opportunities. Science 2000;287:1442–6. expression during expansion and differentiation. Mol Ther 2001;
15. Gage FH. Mammalian neural stem cells. Science 2000;287:1433–8. 3:857–66.
16. Alison M, Sarraf C. Hepatic stem cells. J Hepatol 1998;29:676–82. 37. Pond MJ, Nuki G. Experimentally-induced osteoarthritis in the
17. Murphy JM, Dixon K, Beck S, Fabian DF, Feldman A, Barry FP. dog. Ann Rheum Dis 1973;32:387–8.
Reduced chondrogenic and adipogenic activity of mesenchymal 38. Altman RD, Tenenbaum J, Latta L, Riskin W, Blanco LN, Howell
stem cells from patients with advanced osteoarthritis. Arthritis DS. Biomechanical and biochemical properties of dog cartilage in
Rheum 2002;46:704–13. experimentally induced osteoarthritis. Ann Rheum Dis 1984;43:
18. Creamer P, Hochberg MC. Osteoarthritis. Lancet 1997;350:503–8. 83–90.
19. Buckwalter JA, Mankin HJ. Articular cartilage: degeneration and 39. Rorvik AM, Teige J. Unstable stifles without clinical or radio-
osteoarthritis, repair, regeneration, and transplantation. Instr graphic osteoarthritis in young goats: an experimental study. Acta
Course Lect 1998;47:487–504. Vet Scand 1996;37:265–72.
20. Felson DT, Lawrence RC, Hochberg MC, McAlindon T, Dieppe 40. Little C, Smith S, Ghosh P, Bellenger C. Histomorphological and
PA, Minor MA, et al. Osteoarthritis: new insights. Part 2: treat- immunohistochemical evaluation of joint changes in a model of
ment approaches. Ann Intern Med 2000;133:726–37. osteoarthritis induced by lateral meniscectomy in sheep. J Rheu-
21. Gillquist J, Messner K. Anterior cruciate ligament reconstruction matol 1997;11:2199–209.
and the long-term incidence of gonarthrosis. Sports Med 1999;27: 41. Smith MM, Little CB, Rodgers K, Ghosh P. Animal models used
143–56. for the evaluation of anti-osteoarthritis drugs. Pathol Biol 1997;
22. Wakitani S, Goto T, Pineda SJ, Young RG, Mansour JM, Caplan 45:313–20.
AI, et al. Mesenchymal cell-based repair of large, full-thickness 42. Rangger C, Kathrein A, Klestil T, Glotzer W. Partial meniscec-
defects of articular cartilage. J Bone Joint Surg Am 1994;76: tomy and osteoarthritis: implications for treatment of athletes.
579–92. Sports Med 1997;23:61–8.
23. Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda 43. Gastel JA, Muirhead WR, Lifrak JT, Fadale PD, Hulstyn MJ,
M. Human autologous culture expanded bone marrow mesenchy- Labrador DP. Meniscal tissue regeneration using a collagenous
mal cell transplantation for repair of cartilage defects in osteoar- biomaterial derived from porcine small intestine submucosa. Ar-
thritic knees. Osteoarthritis Cartilage 2002;10:199–206. throscopy 2001;17:151–9.
24. Bruder SP, Jaiswal N, Ricalton NS, Mosca JD, Kraus KH, 44. Peretti GM, Caruso EM, Randolph MA, Zaleske DJ. Meniscal
Kadiyala S. Mesenchymal stem cells in osteobiology and applied repair using engineered tissue. J Orthop Res 2001;19:278–85.
bone regeneration. Clin Orthop 1998;355:S247–56. 45. Rodkey WG, Steadman JR, Li ST. A clinical study of collagen
25. Rydell N, Balazs EA. Effect of intra-articular injection of hyal- meniscus implants to restore the injured meniscus. Clin Orthop
uronic acid on the clinical symptoms of osteoarthritis and on 1999;367:S281–92.
granulation tissue formation. Clin Orthop 1971;80:25–32. 46. Jackson DW, Whelan J, Simon TM. Cell survival after transplan-
26. Kavalkovich KW, Boynton RE, Murphy JM, Barry F. Chondro- tation of fresh meniscal allografts: DNA probe analysis in a goat
genic differentiation of human mesenchymal stem cells within an model. J Sports Med 1993;21:540–50.
alginate layer culture system in vitro. Cell Develop Biol—Animal 47. Rodeo SA. Meniscal allografts: where do we stand? Am J Sports
2002; 38:457–66. Med 2001;29:246–61.
27. Srinivas GR, Chichester CO, Barrach HJ, Pillai V, Matoney AL. 48. Lutfi AM. Morphological changes in the articular cartilage after
Production of type II collagen specific monoclonal antibodies. meniscectomy: an experimental study in the monkey. J Bone Joint
Immunol Invest 1994;23:85–98.
Surg Br 1975;57:525–8.
28. Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-
49. Elmer RM, Moskowitz RW, Frankel VH. Meniscal regeneration
renewal, and the osteogenic potential of purified human mesen-
and postmeniscectomy degenerative joint disease. Clin Orthop
chymal stem cells during extensive subcultivation and following
1977;124:304–10.
cryopreservation. J Cell Biochem 1997;64:278–94.
29. Mosca JD, Hendricks JK, Buyaner D, Davis-Sproul J, Chuang LC, 50. Burr DB, Radin EL. Meniscal function and the importance of
Majumdar MK, et al. Mesenchymal stem cells as vehicles for gene meniscal regeneration in preventing late medical compartment
delivery. Clin Orthop 2000;379:S71–90. osteoarthrosis. Clin Orthop 1982;171:121–6.
30. Keller G, Paige C, Gilboa E, Wagner EF. Expression of a foreign 51. Walsh CJ, Goodman D, Caplan AI, Goldberg VM. Meniscus
gene in myeloid and lymphoid cells derived from multipotent regeneration in a rabbit partial meniscectomy model. Tissue Eng
hematopoietic precursors. Nature 1985;318:149–54. 1999;5:327–37.
31. Ghattas IR, Sanes JR, Majors JE. The encephalomyocarditis virus 52. McClain SA, Simon M, Jones E, Nandi A, Gailit JO, Tonnesen
internal ribosome entry site allows efficient coexpression of two MG, et al. Mesenchymal cell activation is the rate-limiting step of
genes from a recombinant provirus in cultured cells and in granulation tissue induction. Am J Pathol 1996;149:1257–70.
embryos. Mol Cell Biol 1991;11:5848–59. 53. Chen WY, Abatangelo G. Functions of hyaluronan in wound
32. Miller AD, Garcia JV, von Suhr N, Lynch CM, Wilson C, Eiden repair. Wound Repair 1999;7:79–89.
MV. Construction and properties of retrovirus packaging cells 54. Dostal GH, Gamelli RL. Fetal wound healing. Surg Gynecol
based on gibbon ape leukemia virus. J Virol 1991;65:2220–4. Obstet 1993;176:299–306.
33. Markowitz D, Goff S, Bank A. A safe packaging line for gene 55. McCarty MF. Glucosamine for wound healing. Med Hypotheses
transfer: separating viral genes on two different plasmids. J Virol 1996;47:273–5.
1988;62:1120–4. 56. Murashita T, Nakayama Y, Hirano T, Ohashi S. Acceleration of
3474 MURPHY ET AL

granulation tissue ingrowth by hyaluronic acid in artificial skin. cells. Transactions of the 48th Annual Meeting of the Orthopaedic
Br J Plast Surg 1996;49:58–63. Res Society S009; 2002.
57. Kobayashi K, Amiel M, Harwood FL, Healey RM, Sonoda M, 60. Kavalkovich KW, Murphy JM, Barry FP. Adhesion of mesenchy-
Moriya H, et al. The long-term effects of hyaluronan during mal stem cells to fibrillated osteoarthritic cartilage [abstract].
development of osteoarthritis following partial meniscectomy in a Osteoarthritis Cartilage 2000;8 Suppl B:S24.
rabbit model. Osteoarthritis Cartilage 2000:8:359–65. 61. Li B, Aspden RM. Composition and mechanical properties of
58. Brittberg M, Tallheden T, Sjogren-Jansson B, Lindahl A, Peterson cancellous bone from the femoral head of patients with osteo-
L. Autologous chondrocytes used for articular cartilage repair: an porosis or OA. J Bone Miner Res 1997;12:641–51.
update. Clin Orthop 2001;391:S337–48. 62. Dequeker J, Mokassa L, Aerssens J, Boonen S. Bone density and
59. Archer CW, Redman S, Bishop J, Bowyer S, Dowthwaite GP. The local growth factors in generalized OA. Microsc Res Tech 1997;
identification and characterization of articular cartilage progenitor 137:358–71.

Anda mungkin juga menyukai